Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

Embed Size (px)

Citation preview

  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    1/8

    Review article

    Thiazolidinediones for the treatment of type 2 diabetes

    J.W.F. Elte a ,, J.F. Blickl b

    a Sint Franciscus Gasthuis, Department of Internal Medicine, Kleiweg 500, 3045 PM Rotterdam, The Netherlandsb Service de Mdicine Interne, Diabte et Maladies Mtaboliques, Hpitaux Universitaires de Strasbourg, 67091 STRASBOURG Cedex, France

    Received 8 May 2006; received in revised form 1 September 2006; accepted 19 September 2006

    Abstract

    Thiazolidinediones (TZD), or glitazones, represent a new generation of antidiabetic drugs that have recently been introduced in Europe.

    They improve insulin resistance, one of the key anomalies involved in the pathogenesis of type 2 diabetes mellitus, by activating the nuclear

    peroxoxisome proliferator activated receptor- (PPAR-), leading to crucial metabolic alterations in adipose tissue. Rosiglitazone and

    pioglitazone have been shown to be active as monotherapy, in combination therapy with metformin or sulfonylureas, and even in triple

    therapy. They are generally well tolerated but can induce fluid retention. Cardiac failure is a contraindication for the use of TZDs, as is the

    concomitant administration of insulin. Aside from their effect on glycemic control, TZDs act on several cardiovascular risk factors and may

    protect pancreatic cells from apoptosis. The cardiovascular protective effect of TZDs has recently been demonstrated with the results of the

    PROactive study, and long-term preservation of-cell function is currently under further investigation.

    2006 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.

    Keywords: Type 2 diabetes; Thiazolidinediones; Metabolic syndrome; Cardiovascular risk

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

    2. Pharmacological data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

    2.1. Rosiglitazone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

    2.2. Pioglitazone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

    3. Mechanism of action of TZDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

    4. Metabolic effects of TZDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

    4.1. Glucose metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19

    4.2. Lipid metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20

    5. Effects on other cardiovascular risk factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20

    5.1. Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20

    5.2. Microalbuminuria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20

    5.3. Plasminogen activator inhibitor-1 (PAI-1) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205.4. Adipocytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20

    5.5. Fat distribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21

    5.6. Intima-media thickness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21

    5.7. Improvement in cardiovascular risk markers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21

    5.8. Re-stenosis after coronary stent implantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21

    6. Therapeutic perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

    6.1. -cell protection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

    European Journal of Internal Medicine 18 (2007) 1825

    www.elsevier.com/locate/ejim

    Corresponding author. Tel.: +31 104616094; fax: +31 104612692.

    E-mail address:[email protected](J.W.F. Elte).

    0953-6205/$ - see front matter 2006 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.

    doi:10.1016/j.ejim.2006.09.007

    mailto:[email protected]://dx.doi.org/10.1016/j.ejim.2006.09.007http://dx.doi.org/10.1016/j.ejim.2006.09.007mailto:[email protected]
  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    2/8

    6.2. Cardiovascular prevention (PROactive study) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

    7. Adverse effects of TZDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22

    8. TZDs in the treatment of type 2 diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23

    9. Pharmaco-economic evaluation of TZDs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23

    10. Learning points . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23

    1. Introduction

    Insulin resistance plays a major role in the pathogenesis of

    type 2 diabetes [1], a disease leading to severe long-term

    cardiovascular complications. The glycemic deterioration

    observed over time in type 2 diabetes is attributable to a

    progressive decline in -cell function[2]. Control of blood

    glucose levels is essential for the prevention of complications

    of the disease[3,4]. Metformin, an insulin sensitizer acting

    predominantly on the liver by reducing hepatic glucose pro-duction, and sulfonylureas, which stimulate insulin release,

    rarely allow for long-term glycemic normalization, even

    when used in combination therapy, since they do not slow -

    cell apoptosis.

    The recently introduced thiazolidinediones (TZDs),

    which act predominantly by enhancing peripheral insulin

    sensitivity, offer promising perspectives in terms of-cell

    preservation[5,6]and cardiovascular protection[7,8].

    2. Pharmacological data

    2.1. Rosiglitazone

    After oral administration of 2 mg rosiglitazone, the

    maximal concentration (Cmax) is achieved after 1.3 h, and

    the elimination half-life of the drug is 3.6 h. Food intake

    slightly slows the rate of absorption of the drug but not the

    amount of drug absorbed. The pharmacokinetics of rosigli-

    tazone is not altered by age or mild to moderate renal

    impairment, but hepatic dysfunction significantly increases

    the area under the concentration curve. Rosiglitazone does

    not induce cytochrome P 450 3A4 metabolism. No drug

    interactions have been observed with ranitidine, metformin,

    or digoxin, but co-administration with acarbose slightlyreduces the absorption of rosiglitazone[9].

    2.2. Pioglitazone

    The time to Cmax and the elimination half-life of

    pioglitazone are slightly longer than for rosiglitazone. The

    drug undergoes extensive hepatic metabolism via the CYP

    2C8 and, more accessorily, the CYP 3A4, 2C9, and 1A1/2.

    Some metabolites (MII, III, IV) are active. Renal impairment

    leads to increased hepatic clearance by reduction of protein

    binding of the drug but does not alter the free plasma drug

    concentrations. No induction or inhibition of hepatic enzyme

    systems and no clinically significant drug interactions have

    been reported to date with pioglitazone[10].

    3. Mechanism of action of TZDs

    It was shown soon after their discovery that TZDs are

    agonists of the peroxysome proliferator activated receptors-

    (PPAR-) [11,12]. Briefly, after the binding of a TZD to

    PPAR-, the macromolecular complex formed by PPAR-

    and the retinoic acid receptor is able to recruit an activator thatallows the DNA transcription of peroxysome proliferator

    response elements (PPRE; Fig. 1). PPAR- is essentially

    expressed in adipose tissue and controls genes that are mostly

    involved in adipocyte differentiation and lipid metabolism.

    This cannot entirely explain the glucose-lowering effect of

    the drug since adipose tissue accounts for less than 5% of

    glucose utilization. The explanation of this paradox is that

    TZDs promote the differentiation of adipose tissue into small

    adipocytes, which are more insulin-sensitive than large adi-

    pocytes and, therefore, release into the bloodstream fewer

    free fatty acids (FFA), more adiponectin, and less TNF-,

    resistin, and leptin. This leads to an improvement in peri-

    pheral glucose uptake in the skeletal muscle, a decrease inhepatic glucose production, and an increase in fat storage in

    adipose tissue[13].

    4. Metabolic effects of TZDs

    4.1. Glucose metabolism

    In placebo-controlled studies, TZDs decrease fasting

    plasma glucose and HbA1c levels in a dose-dependent

    manner in monotherapy as well as in combination with met-

    formin, sulfonylureas, or even in triple therapy[9,10]. This

    Fig. 1. Mechanism of action of TZDs.

    19J.W.F. Elte, J.F. Blickl / European Journal of Internal Medicine 18 (2007) 1825

  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    3/8

    glucose-lowering effect is related to an improvement in

    insulin sensitivity, as suggested by a decrease in plasma

    insulin levels during TZD treatment.

    In comparison to glibenclamide or gliclazide, the decrease

    in FPG and HbA1c is slower with TZDs and the maximal

    effect is reached only after 12 weeks, in accordance with the

    indirect mechanism of action of these drugs. By contrast,secondary deterioration of glycemic control is faster with

    sulfonylurea treatment than with TZD treatment [14]. The

    same is true to a lesser extent for comparative studies of

    TZDs with metformin[15].

    4.2. Lipid metabolism

    Both TZDs greatly decrease FFA levels and significantly

    increase HDL-C levels. Their effect on triglycerides and LDL-

    C levels is, however, different, perhaps because of a higher

    PPAR- selectivity of rosiglitazone. The differences observed

    in several placebo-controlled trials and switch studies fromtroglitazone to rosiglitazone or pioglitazone [16]have been

    confirmed recently in a head-to-head comparative study

    showing a decrease in fasting triglycerides and postprandial

    lipemia[17]and stability of LDL-C with pioglitazone, a non-

    significant variation of triglycerides, and an increase in LDL-C

    with rosiglitazone[18]. Both drugs reduce the proportion of

    small dense atherogenic LDL particles, but pioglitazone does

    so more efficiently than rosiglitazone.

    5. Effects on other cardiovascular risk factors

    5.1. Hypertension

    Most of the studies investigating the effects of TZDs on

    blood pressure have been performed with rosiglitazone and

    had a rather short duration. Also, the patient populations

    studied were different, i.e., hypertensive patients [19],

    patients with impaired glucose tolerance [20], and type 2

    diabetes patients with [21] or without [22] hypertension.

    These studies were most often open-label and not always

    controlled and/or randomized. However, all studies showed

    positive effects on ambulatory systolic and diastolic

    pressure. The study with the longest duration was the

    open-label, active controlled study by Sutton et al. [22], who

    showed a significant decrease in diastolic blood pressure(2.3 mm Hg) after 52 weeks of 4 mg rosiglitazone (whereas

    in the glibenclamide group there was no difference). Systolic

    blood pressure did not change during rosiglitazone therapy,

    but it increased significantly in the glibenclamide patients

    (+3.8 mm Hg). There were no changes in left ventricular

    mass or ejection fraction in either group.

    5.2. Microalbuminuria

    Microalbuminuria may be considered a risk indicator of

    vascular damage in type 2 diabetes. Therefore, a reduction

    in microalbuminuria reflecting a decrease in vascular

    risk may be beneficial. Bakris et al. [23] performed a

    52-week, open-label, randomized study comparing the

    effects of rosiglitazone (4 mg) and glibenclamide (mean

    dose 10.5 mg) on microalbuminuria. After 28 weeks, a

    significant reduction in the albumin/creatinine ratio (ACR)

    was observed in both treatment groups, whereas after

    52 weeks this was only true in the rosiglitazone group(normalization in 43% versus 6% in the glibenclamide

    group). Not unexpectedly, there was a strong correlation

    of the ACR reduction with changes in mean 24-hour

    systolic and diastolic blood pressure in the rosiglitazone

    group (not in the glibenclamide patients), but not with the

    glucose metabolism parameters. Similar observations have

    been found in an earlier double-blind, placebo-controlled

    study of rosiglitazone [24].

    5.3. Plasminogen activator inhibitor-1 (PAI-1)

    One of the components of the metabolic syndrome isincreased PAI-1, which is associated with cardiovascular

    risk, probably because PAI-1 is associated with deficient

    fibrinolysis and intravascular thrombosis. Preliminary results

    of a double-blind, randomized, parallel-group study com-

    paring single drug treatment with glibenclamide with the

    combination of glibenclamide (up to 10 mg/day) plus 4 mg

    rosiglitazone twice daily for 26 weeks were favorable[25].

    PAI-1 activity, PAI-1 antigen, and tPA decreased signifi-

    cantly (33.8, 21.8, and 25.3%, respectively) in the group

    using rosiglitazone compared to the group only using

    glibenclamide. A significant decrease was also observed

    after 26 weeks of rosiglitazone plus glibenclamide with

    respect to baseline values, something that may contribute tothe beneficial effect of rosiglitazone in endothelial dysfunc-

    tion and to the decrease in cardiovascular complications[25].

    5.4. Adipocytokines

    Adipocytes (or fat cells) contain PPAR- in high

    concentration and also secrete many substances with

    metabolic activity, the so-called adipocytokines. These

    include substances with beneficial (e.g., leptin, adiponectin)

    as well as disadvantageous effects (e.g., free fatty acids

    (FFA), TNF, IL-6, PAI-1, and resistin).

    In a double-blind, placebo-controlled study of 23 type 2diabetes patients already treated with sulfonylurea drugs,

    Miyazaki et al. [26] investigated the effect of 45 mg

    pioglitazone (n =12) versus placebo (n =11) for 4 months on

    various adipocytokines. Apart from a decrease in HbA1c and

    fasting plasma glucose, pioglitazone showed significant

    decreases in circulating concentrations of FFA and TNF

    and increments of adiponectin in comparison with baseline

    and placebo. Plasma leptin did not change significantly in

    either group. These direct effects of pioglitazone may very

    well contribute to the improved hepatic and peripheral

    insulin sensitivity and ameliorate glucose tolerance in type 2

    diabetic patients.

    20 J.W.F. Elte, J.F. Blickl / European Journal of Internal Medicine 18 (2007) 1825

  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    4/8

    In a study on non-traditional risk factors for cardiovas-

    cular disease in type 2 diabetes patients, Haffner et al. [27]

    showed that rosiglitazone reduced levels of matrix metallo-

    proteinase-9 (MMP-9) and C-reactive protein (CRP), but not

    of IL-6 and white blood cell count after 26 weeks of

    treatment. These data may explain in part the beneficial

    effects of rosiglitazone on cardiovascular risk. A summary ofthe effects of TZDs on cardiovascular risk factors is

    presented inTable 1.

    5.5. Fat distribution

    The most prominent feature and probable cause of the

    metabolic syndrome and of (hepatic) insulin resistance is

    visceral fat accumulation. TZDs can cause a shift in fat

    distribution from visceral to subcutaneous depots [28,29].

    Although the total fat mass increases, HbA1c decreases due

    to improved hepatic and peripheral tissue insulin sensitivity.

    The concomitant decreased FFA plasma concentration pointsto a healthier fat cell, responding now to insulin stimulation

    of glucose uptake and suppression of lipolysis[29].

    5.6. Intima-media thickness

    Intima-media thickness (IMT), measured with the help of

    ultrasound, has been used as a surrogate marker of early

    atherosclerotic lesions, and IMT may be considered a

    surrogate endpoint of atherosclerotic disease. The effect of

    TZDs on IMT progression has been reported in studies with

    approximately 50170 patients for a duration of 6

    24 months [3034]. The studies were mostly open; only

    one was randomized and double-blind [32]. Troglitazone[30], rosiglitazone (for non-diabetics with coronary artery

    disease)[32], and pioglitazone treatment[31,33,34]resulted

    in a significant decrease in IMT, pointing to a reduction in

    atherosclerosis in diabetic and non-diabetic patients.

    5.7. Improvement in cardiovascular risk markers

    In a 6-month, prospective, open-label, controlled clinical

    study with 192 patients, Pftzner et al. [35] examined the

    effects of pioglitazone on inflammatory and atherogenic

    markers, both biochemical and clinical, as compared with

    glimepiride. Although HbA1c reduction was comparable inboth groups, most parameters improvedmore effectively in the

    pioglitazone-treated patients. These parameters included

    insulin, LDL-C/HDL-C ratio, high-sensitivity CRP, MMP-9,

    MCP-1 (monocyte chemoattractant protein-1), and carotid

    IMT. No changes were seen in LDL-cholesterol, triglycerides,

    fibrinogen, von Willebrand factor, PAI-1, and a number of

    other markers of endothelial (dys)function. It may be

    concluded that pioglitazone has anti-inflammatory and anti-

    atherogenic effects, independent of blood glucose control.

    5.8. Re-stenosis after coronary stent implantation

    Coronary stent implantation leads to a high rate of re-

    stenosis, especially in diabetics, resulting in a poorer long-

    term prognosis than in non-diabetics. Antiplatelet drugs,

    anticoagulants, and statins have not been successful in

    reducing re-stenosis [36]. PPAR-g agonists inhibit the

    growth of vascular smooth muscle cells (VSMC) and may

    prevent neo-intima formation and re-stenosis.

    Apart from one small study with negative results

    concerning 16 diabetes type 2 patients (8 on rosiglitazone

    and 8 on placebo) [37], there are now three randomized

    studies [3839] showing a decrease in re-stenosis after

    6 months of treatment with either rosiglitazone [35,38] or

    pioglitazone[38].Choi et al. investigated 83 type 2 diabetes patients in a

    prospective, randomized, case-controlled trial and found a

    more than 50% reduction in the occurrence of re-stenosis and

    a lower degree of stenosis of the luminal diameter after

    rosiglitazone. High-sensitivity C-reactive protein (CRP) was

    reduced, but glucose and lipid parameters remained

    unchanged[36].

    Wang et al. studied 71 randomly divided patients

    (rosiglitazone versus placebo) [39]. Plasma monocyte

    chemoattractant protein-1 (MCP-1) and CRP decreased,

    but fasting glucose, insulin, and HbA1c were significantly

    lowered in the rosiglitazone group. The occurrence ofcoronary events decreased, probably by not only improving

    metabolic parameters but also by reducing inflammatory

    responses[39].

    Finally, Marx et al. performed a randomized, placebo-

    controlled, double-blind trial with pioglitazone in 50 non-

    diabetic CAD patients. Fibrinogen levels decreased signif-

    icantly, but CRP, TNF-, glucose parameters, and lipids did

    not. Neo-intima volume, total plaque volume, and mean

    stenosis of the luminal diameter decreased, suggesting a

    direct effect of TZD treatment on neo-intima volume after

    coronary stent implantation [38], independent of its

    metabolic actions.

    Table 1

    Effects of glitazone therapy on established and emerging CVD risk factors

    (adapted from[61])

    CVD risk factor Impact of glitazone therapy

    Hyperglycemia Reduction in HbA1c

    Hypertension Reduction in blood pressure

    Dyslipidemia Reduction in triglyceridesIncrease in HDL cholesterol

    Increase in LDL particle size (fewer

    atherogenic particles)

    Decrease in FFA

    Decrease in postprandial lipemia

    Markers of endothelial

    inflammation

    Decreased C-reactive protein

    Decreased white blood cell count

    Decreased fibrinogen

    Decreased matrix metalloproteinase-9

    Increased adiponectin

    Decreased tumor necrosis factor-

    Decreased microalbuminuria

    Markers of elevated

    thrombotic risk

    Decreased plasminogen activator inhibitor-1

    Decreased platelet aggregation

    21J.W.F. Elte, J.F. Blickl / European Journal of Internal Medicine 18 (2007) 1825

  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    5/8

    6. Therapeutic perspectives

    6.1. -cell protection

    It has been well established in rats [40] and mice [41]

    that TZDs may prevent the progression from insulin

    resistance to overt diabetes by preserving -cell mass andinsulin secretion capacity. In type 2 diabetics, rosiglitazone,

    as compared to placebo or glibenclamide, significantly

    reduced the proinsulin/insulin ratio after 26 and 52 weeks,

    which is compatible with a reduction in -cell dysfunction

    [42]. Moreover, pioglitazone significantly improved -cell

    response by HOMA assessment when used either as

    monotherapy or in combination with metformin or sul-

    fonylurea for a period of 16 or 26 weeks [24,43,44]. In a

    placebo-controlled, 26-week study of pioglitazone (30

    45 mg), Miyazaki et al. [45]showed an increase in plasma

    insulin response during OGTT in diabetic patients. At the

    same time, plasma glucose levels decreased, pointing to animprovement in -cell function. Juhl et al. [46]concluded

    from their study that 3 months of rosiglitazone treatment in

    type 2 diabetics did not influence insulin secretion per se,

    but that improved glucose-entrained, high-frequency insu-

    lin pulsatility suggested an increased ability of the cell to

    sense and respond to glucose changes within the physio-

    logical range.

    TZDs may improve -cell function through several

    mechanisms[47]. Improved insulin sensitivity may reduce

    glucotoxicity of the cell, but lipotoxicity may decrease as

    well, as a result of the reduction in circulating FFA caused by

    increased insulin sensitivity of the cell and reduced levels of

    TNF[47]. In animals it has been shown that TZDs preventdeterioration of islet cell morphology, preserving pancreatic

    content and -cell ultrastructure[40].

    6.2. Cardiovascular prevention (PROactive study)

    The results of the PROactive study have recently been

    reported[8]. This prospective, randomized, controlled study

    included 5238 type 2 diabetes patients with macrovascular

    complications who were followed for 3.45 years. Pioglita-

    zone was given in addition to existing therapies. The

    expectations were high, but the results were not easy to

    interpret and have caused a lot of debate [4850].Some issues should be highlighted. The primary endpoint

    was a composite of disease endpoints (death, myocardial

    infarction, stroke, acute coronary syndrome), but also

    procedural endpoints (coronary and leg revasculations, leg

    amputations). Probably because of the inclusion of the

    procedural endpoints, any advantage that pioglitazone may

    have had over placebo could not be confirmed as far as the

    primary endpoint was concerned. The secondary endpoint

    consisted of the separate diseases of the primary endpoint,

    i.e., myocardial infarction, stroke, and (cardiovascular)

    death. Here, a significant decrease of 16% (p =0.027) was

    observed in the pioglitazone group[8].

    Other problems included the rather fast recruitment and

    closure, possibly decreasing the power of the study. Also,

    one-third of the patients were using insulin, and it is not clear

    from the study whether these were the patients with the

    highest risk of heart failure/edema. There was a significant

    increase in edema not attributable to heart failure (221 events

    more in the pioglitazone group) as well as in heart failure(115 events more, p =b0.001), but no excess mortality.

    Although more hypoglycemias occurred during pioglitazone

    treatment, the hospital admission rate remained the same.

    There were more pneumonias in the pioglitazone group and a

    weight gain of 4 kg compared to the placebo group. A

    positive result was that insulin treatment could be reduced

    (or postponed) by 50% with pioglitazone[8].

    Thus, pioglitazone reduced cardiovascular morbidity and

    mortality (but only measured with the secondary endpoint) in

    type 2 diabetics with a high risk of macrovascular

    complications. It reduced the need for insulin treatment,

    but caused weight gain, edema, and heart failure[8].It is still not known which patients are at the greatest risk

    of heart failure after treatment with TZDs, what the

    prognosis of heart failure is, and whether it is safe to

    combine insulin and pioglitazone treatment[48]. InTable 2

    an overview is given of proven and potential benefits and

    risks of TZDs.

    7. Adverse effects of TZDs

    TZDs do not induce hypoglycemias in monotherapy, but

    they do slightly increase the risk in combination with

    sulfonylureas.

    With pioglitazone and rosiglitazone, no cases of severehepatotoxicity, such as those which led to the withdrawal of

    troglitazone from the market, have been observed[51], and

    in long-term cohort studies, like the PROactive study, TZDs

    decreased ALT levels by improving liver steatosis [8].

    Table 2

    Proven and potential benefits and risks of thiazolidinediones (adapted from

    [62])

    Benefits

    Improved glycemic control

    Lower insulin resistance/insulin levels

    Fat redistribution/decreased visceral fatLower blood pressure

    Improved pancreatic -cell function

    Improved endothelial function/decreased IMT

    Reduced cardiovascular morbidity and mortality (PROactive)

    Induction of ovulation in PCOS

    Less bone turnover

    Treatment for neoplasms

    Decreased ALAT, suggesting decreased liver fat

    Risks

    Hepatotoxicity/potential for liver failure

    Weight gain/increased total body fat (subcutaneous)

    Edema/fluid retention

    Pulmonary edema

    Increased Lp(a) lipoprotein levels

    22 J.W.F. Elte, J.F. Blickl / European Journal of Internal Medicine 18 (2007) 1825

  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    6/8

    The main adverse effects of TZDs are related to fluid

    retention[52], which can result in pseudo-anemia, edema,

    and cardiac failure in patients with underlying heart disease.

    This has resulted in the restricted use of TZDs in Europe.

    However, TZDs do not induce cardiac hypertrophy or reduce

    the cardiac ejection fraction. The mechanism underlying the

    edema is unclear but probably involves both fluid retentionand increased vascular permeability. In the majority of cases,

    edema is not related to cardiac insufficiency.

    Another frequent, and possibly limiting, adverse effect is

    weight gain, which is a consequence of the mechanism of

    action of TZDs. The average increase in body weight is

    about 23 kg, but it can be much more in some subjects. It

    generally occurs during the first year of treatment with no

    further increment. It has been shown to be related to the

    development of subcutaneous fat with no significant mod-

    ification or even a trend to a decrease in abdominal fat and, as

    a consequence, no increase in insulin resistance or loss of

    therapeutic efficacy[53].

    8. TZDs in the treatment of type 2 diabetes

    The classical approach to the treatment of type 2 diabetes

    is stepwise, starting with diet and exercise, followed by the

    initiation of oral monotherapy, leading after a few years to

    combination therapy and, finally, to insulin therapy [54].

    The change in therapy is generally dictated by the manifest

    failure of the treatment, with the mean glycemic control,

    i.e., HbA1c and glucose levels, over the years being above

    the recommended target goals. In many countries, sulfony-

    lureas are frequently used as first-line drugs, and the dose is

    increased to the maximum recommended dose beforestarting combination therapy. In fact, in the majority of

    cases, an insulin sensitizer probably represents a better

    choice for the initial drug therapy. Metformin has been the

    recommended first-line drug ever since the UKPDS showed

    a benefit of this drug versus sulfonylureas or insulin in the

    prevention of cardiovascular events in overweight type 2

    diabetic patients. In the case of a contraindication or in-

    tolerance to metformin, a TZD can be used alternatively

    with the goal of attaining a normal or near-normal HbA1c

    level, which can be targeted because insulin sensitizers do

    not induce severe hypoglycemia. The combination with a

    TZD represents a good therapeutic option if HbA1c exceeds6.5% under a maximal, tolerated metformin regimen in

    obese or overweight patients.

    In lean subjects, the addition of a TZD to sulfonylurea

    monotherapy is allowed only if metformin is contraindicated

    or not tolerated.

    Finally, rosiglitazone can now be used in triple therapy

    with metformin and sulfonylurea. This strategy represents a

    valid alternative to basal insulin therapy combined with oral

    antidiabetic drugs, particularly in the 79% HbA1c range

    [55]. In more severely decompensated patients, insulin

    should be preferred and, in that case, the TZD treatment

    withdrawn.

    9. Pharmaco-economic evaluation of TZDs

    The cost-effectiveness analyses performed to date have

    mainly been based on several combination therapy trials with

    an extrapolation of the event rates according to the UKPDS

    model with some local adaptations. It has been concluded

    that, in overweight type 2 diabetics, combined therapy withpioglitazone increases life expectancy at an acceptable cost

    for Germany[56]. In Sweden, the cost per life-year gained

    with a combination of pioglitazone and metformin or sulfo-

    nylurea is comparable with current treatments and can be

    considered as cost-effective in the national health care system

    [57]. The evaluation of pioglitazone in comparison to other

    strategies as first-line treatment in Canada also concluded

    that, for certain patient strata, this therapeutic alternative

    could be cost-effective[58]. In comparison to insulin, TZD

    treatment with rosiglitazone or pioglitazone reduced diabe-

    tes-related costs despite higher diabetes-related pharmacy

    costs in the US[59].More precise pharmaco-economic evaluations will be

    available after the publication of long-term trials, like

    DREAM and RECORD. Based on the only available

    endpoint study to date, PROactive, an economic evaluation

    of pioglitazone on therapy in a predefined approach is being

    planned in order to determine the cost per life-year gained in

    every country and to help improve the allocation of health

    care resources[60].

    10. Learning points

    Thiazolidinediones (TZD) improve insulin resistance.

    Fluid retention and weight gain may occur. Cardiovascular risk factors, including hypertension,

    lipids, microalbuminuria, PAI-1, endothelial function,

    and IMT improve after TZD therapy.

    TZDs cause fat redistribution from visceral to

    subcutaneous depots. They reduce adipocytokines

    and, most likely, liver fat content.

    TZDs improve -cell function.

    Pioglitazone reduces cardiovascular morbidity and

    mortality in high-risk diabetes 2 patients (PROactive).

    References

    [1] Reaven GM. Role of insulin resistance in human disease. Diabetes

    1988;37:1595607.

    [2] UK prospective diabetes study (UKPDS) group. Overview of

    6 years therapy of type 2 diabetes: a progressive disease. Diabetes

    1995;44:124950.

    [3] UK prospective diabetes study (UKPDS) group. Intensive blood-

    glucose control with sulfonylureas or insulin compared with

    conventional treatment and risk of complications in patients with

    type 2 diabetes (UKPDS 33). Lancet 1998;352:83753.

    [4] UK prospective diabetes study (UKPDS) group. Effect of intensive

    blood-glucose control with metformin on complications in overweight

    patients with type 2 diabetes (UKPDS 34). Lancet 1998;352:85465.

    [5] Buchanan TA, Xiang AH, Peters RK, Kjos SL, Marroquin A, Goico J,

    et al. Preservation of pancreatic beta-cell function and prevention of

    23J.W.F. Elte, J.F. Blickl / European Journal of Internal Medicine 18 (2007) 1825

  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    7/8

    type 2 diabetes by pharmacological treatment of insulin resistance in

    high-risk Hispanic women. Diabetes 2002;51:2796803.

    [6] Ovalle F, Bell DSH. Clinical evidence of thiazolidinedione-induced

    improvement of pancreatic -cell function in patients with type 2

    diabetes mellitus. Diabetes Obes Metab 2002;4:569.

    [7] Suwattee P, DeSouza C, Asnani S, GillingL, Fonseca VA. Cardiovascular

    effects of thiazolidinediones. Endocrinologist 2002;12:12634.

    [8] Dormandy JA, Charbonnel B, EcklandDJA, ErdmannE, Massi-BenettiM, Moules IK, et al. Secondary prevention of macrovascular events in

    patients with type 2 diabetes in the PROactive Study (PROspective

    pioglitAzone Clinical Trial In macroVascular Events): a randomized

    controlled trial. Lancet 2005;366:127989.

    [9] Barman-Balfour JA, Plosker GL. Rosiglitazone. Drugs1999;57:92130.

    [10] Gillies PS, Dunn CJ. Pioglitazone. Drugs 2000;60:33343.

    [11] Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson

    TM, Kliewer SA. An antidiabetic thiazolidinedione is a high affinity

    ligand for peroxisome proliferators-activated receptor (PPAR).

    J Biol Chem 1995;270:129536.

    [12] Vamecq J, Latruffe N. Peroxisome proliferators-activated receptors

    (PPARs) and their implications in diseases. Curr Opin Endocrinol

    Diabetes 2000;7:818.

    [13] Tan MH. How pioglitazone affects glucose and lipid metabolism. Exp

    Clin Endocrinol Diabetes 2000;108(suppl 2):S22433.[14] Tan MH, Baksi A, Krahulec B, Kubalski P, Stankiewicz A, Urquhart R,

    et al. Comparison of pioglitazone and gliclazide in sustaining glycemic

    control over 2 years in patients with type 2 diabetes. Diabetes Care

    2005;28:54450.

    [15] Hanefeld M, et al, on behalf of the QUARTET Study Group. One-year

    glycemic control with a sulfonylurea plus pioglitazone versus a

    sulfonylurea plus metformin in patients with type 2 diabetes. Diabetes

    Care 2004;27:1417.

    [16] Khan MA, St. Peter JV, Xue JL. A prospective, randomized

    comparison of the metabolic effects of pioglitazone or rosiglitazone

    in patients with type 2 diabetes who were previously treated with

    troglitazone. Diabetes Care 2002;25:70811.

    [17] van Wijk JPH, de Koning EJP, Castro Cabezas M, Rabelink TJ.

    Rosiglitazone improves postprandial triglyceride and free fatty acid

    metabolism in type 2 diabetes. Diabetes Care 2005;28:8449.[18] Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, Pinaire JA,

    et al. A comparison of lipid and glycemic effects of pioglitazone and

    rosiglitazone in patients with type 2 diabetes and dyslipidemia.

    Diabetes Care 2005;28:154754.

    [19] Raji A, Seely EW, Bekins SA, Williams GH, Simonson DC.

    Rosiglitazone improves insulin sensitivity and lowers blood pressure

    in hypertensive patients. Diabetes Care 2003;26:1728.

    [20] Bennett SMA, Agrawal A, Elashat H. Rosiglitazone improves

    insulin sensitivity, glucose tolerance and ambulatory blood pressure

    in subjects with impaired glucose tolerance. Diabet Med

    2004;21:41522.

    [21] Sarafidis PA, Lasaridis AN, Nilsson PM, Pagkalos EM, Hitoglou-

    Makedou AD, Pliakos CI, et al. Ambulatory blood pressure reduction

    after rosiglitazone treatment in patients with type 2 diabetes and

    hypertension correlates with insulin sensitivity increase. J Hypertens2004;22:176977.

    [22] St John Sutton MJ, Rendell M, Dandona P, Dole JF, Murphy K,

    Patwardhan R, et al. A comparison of the effects of rosiglitazone and

    glyburide on cardiovascular function and glycemic control in patients

    with type 2 diabetes. Diabetes Care 2002;25:205864.

    [23] Bakris G, Viberti G, Weston WM, Heise M, Porter LE, Freed MI.

    Rosiglitazone reduces urinary albumin excretion in type II diabetes.

    J Hum Hypertens 2003;17:712.

    [24] Lebovitz HE, Dole JF, Patwardhan R, Rappaport EB, Freed MI, for the

    rosiglitazone clinical trials study group. Rosiglitazone monotherapy is

    effective in patients with type 2 diabetes. J Clin Endocrinol Metab

    2001;86:2808.

    [25] Freed M, Fuell D, Menci L, Heise M, Goldstein B. Effect of

    combination therapy with rosiglitazone and glibenclamide on PAI-

    1 antigen, PAI-1 activity, and tPA in patients with type 2 diabetes

    (Abstract). Diabetologia 2000;43(Suppl 1):A267 poster 1024.

    [26] Miyazaki Y, Mahankali A, Wajcberg E, Bajaj M, Mandarino LJ,

    DeFronzo RA. Effects of pioglitazone on circulating adipocytokine

    levels and insulin sensitivity in Type 2 diabetic patients. J Clin

    Endocrinol Metab 2004;89:43129.

    [27] Haffner SM, Greenberg AS, Weston WM, Chen H, Williams K,

    Fred MI. Effect of rosiglitazone treatment on nontraditionalmarkers of cardiovascular disease in patients with type 2 diabetes

    mellitus. Circulation 2002;106:67984.

    [28] Miyazaki Y, Mahankali A, Matsuda M, Mahankali S, Hardies J, Cusi K,

    et al. Effect of pioglitazone on abdominal fat distribution and insulin

    sensitivity in type 2 diabetic patients. J Clin Endocrinol Metab

    2002;87:278491.

    [29] Bays H, Mandarino L, DeFronzo R. Role of the adipocyte, free fatty

    acids, and ectopic fat in pathogenesis of type 2 diabetes mellitus:

    peroxisomal proliferator-activated receptor agonists provide a rational

    therapeutic approach. J Clin Endocrinol Metab 2004;89:46378.

    [30] Minamikawa J, Tanaka S, Yamauchi M, Inoue D, Koshiyama H. Potent

    inhibitory effect of troglitazone on carotid arterial wall thickness in

    type 2 diabetes. J Clin Endocrinol Metab 1998;83:181820.

    [31] Koshiyama H, Shimono D, Kuwamura N, Minamikawa J, Nakamura

    Y. Inhibitory effect of pioglitazone on carotid arterial wall thickness intype 2 diabetes. J Clin Endocrinol Metab 2001;86:34526.

    [32] Sidhu JS, Kaposzta Z, Markus HS, Kaski JC. Effect of rosiglitazone on

    common carotid intima-media thickness progression in coronary artery

    disease patients without diabetes mellitus. Arterioscler Thromb Vasc

    Biol 2004;24:9304.

    [33] NakamuraT, Matsuda T, Kawagoe Y, Ogawa H, Takahashi Y, Sekizuka

    K, et al. Effect of pioglitazone on carotid intima-media thickness and

    arterial stiffness in type 2 diabetic nephropathy patients. Metabolism

    2004;53:13826.

    [34] Langenfeld MR, Forst T, Hohnberg C, Kann P, Lbben G, Konrad T,

    et al. Pioglitazone decreases carotid intima-media thickness indepen-

    dently of glycemiccontrolin patients withtype 2 diabetesmellitus. Results

    from a controlled randomized study. Circulation 2005;111:252531.

    [35] PftznerA, MarxN, LbbenG, Langenfeld M, Walcher D, Konrad T, et al.

    Improvementof cardiovascular risk markers by pioglitazone is independentfrom glycemiuc control. J Am Coll Cardiol 2005;45:192531.

    [36] Choi D, Kim S-K, Choi S-H, Ko YG, Ahn CW, Jang Y, et al.

    Preventative effects of rosiglitazone on restenosis after coronary stent

    implantation in patients with type 2 diabetes. Diabetes Care

    2004;27:265460.

    [37] Osman A, Otero J, BrizolaraA, Waxman S, Stouffer G, FitzgeraldP,et al.

    Effect of rosiglitazone on restenosis after coronary stenting in patients

    with type 2 diabetes. Am Heart J 2004;147:215.

    [38] Marx N, Whrle JH, Nusser T, Walcher D, Rinker A, Hombach V, et al.

    Pioglitazone reduces neointima volume after coronary stent implan-

    tation. A randomized, placebo-controlled, double-blind trial in non-

    diabetic patients. Circulation 2005;112:27928.

    [39] Wang GM, Wei J, Guan Y, Jin N, Mao J, Wang X. Peroxisome

    proliferator-activated receptor-y agonist rosiglitazone reduces clinical

    inflammatory responses in type 2 diabetes with coronary artery diseaseafter coronary angioplasty. Metab Clin Exp 2005;54:5907.

    [40] Smith SA, Lister CA, Toseland CDN, Buckingham RE. Rosiglitazone

    prevents the onset of hyperglycaemia and proteinuria in the Zucker

    diabetic fatty rat. Diabetes Obes Metab 2000;2:36372.

    [41] Ozawa S, Takizawa M, Itoh E, Katsuta H, Tanaka T, Yamaguchi S,

    et al. The protective effect of the long-term treatment by pioglitazone

    on the insulin secretory capacity and cell mass in obese diabetic

    db/db mice. Diabetes 2002;51(suppl 2):A 361 poster 1476-P.

    [42] Porter LE, Freed MI, Jones NP. Rosiglitazone reduces proinsulin/

    insulin ratio and improves -cell function in type 2 diabetes.

    Diabetologia 2000;43(suppl 1):A192 poster 737.

    [43] Rosenstock J, for the pioglitazone HCI study group. Improved insulin

    sensitivity and beta cell responsivity suggested by homa analysis of

    pioglitazone therapy. Diabetologia 2000;43(suppl 1):A192 poster 738.

    24 J.W.F. Elte, J.F. Blickl / European Journal of Internal Medicine 18 (2007) 1825

  • 8/14/2019 Thiazolidinediones for the Treatment of Type 2 Diabetes EJIM 2007

    8/8

    [44] Matthews DR. Insulin resistance and -cell function a clinical

    perspective. Diabetes Obes Metab 2001;3(suppl 1):S2833.

    [45] Miyazaki Y, Matsuda M, DeFronzo RA. Doseresponse effect of

    pioglitazone on insulin sensitivity and insulin secretion in type 2

    diabetes. Diabetes Care 2002;25:51723.

    [46] Juhl CB, Hollingdal M, Prksen N, Prange A, Lnnqvist F, Schmitz O.

    Influence of rosiglitazone treatment on-cell function in type 2 diabetes:

    evidence of an increased ability of glucose to entrain high-frequencyinsulin pulsatility. J Clin Endocrinol Metab 2003;88(8):3794800.

    [47] Leiter LA. -cell preservation: a potential role for thiazolidinediones to

    improve clinical care in type 2 diabetes. Diabet Med 2005;22:96372.

    [48] Yki-Jrvinen H. The PROactive study: some answers, many questions.

    Lancet 2005;366:12412.

    [49] Fonseca V, Jawa A, Asnani S. Commentary: the PROactive study

    the glass is half full. J Clin Endocrinol Metab 2006;91:257.

    [50] Ceriello A. PROactive study : (r)evolution in the therapy of diabetes ?

    Diabet Med 2005;22:14634.

    [51] Scheen AJ. Hepatotoxicity with thiazolidinediones. Is it a class effect?

    Drug Safety 2001;24:87388.

    [52] NestoRW, Bell D, Bonow RO, Fonseca V, Grundy SM, HortonES, et al.

    Thiazolidinedione use, fluid retention, and congestive heart failure.

    Circulation 2003;108:29418.

    [53] Nakamura T, Funahashi T, Yamashita S, Nishida M, Nishida Y,Takahashi M, et al. Thiazolidinedione derivative improves fat distri-

    bution and multip le risk factors in subject s with viscer al fat

    accumulation-double-blind placebo-controlled trial. Diabetes Res

    Clin Pract 2001;54:18190.

    [54] IDF. A desktop guide to type 2 diabetes. Diabet Med 1999;16:71630.

    [55] Rosenstock J, Sugimoto D, Strange P, Stewart J, Soltes-Rak E, Dailey

    G. Triple therapy in type 2 diabetes (T2DM): benefits of insulin

    glargine (GLAR) over rosiglitazone (RSG) added to combination

    therapy of sulfonylurea plus metformin (SU+MET) in insulin-nave

    patients. Diabetes 2004(suppl 3) poster 609.

    [56] Neeser K, Lbben G, Siebert U, Schramm W. Cost effectiveness of

    combination therapy with Pioglitazone for type 2 diabetes mellitus

    from a German statutory healthcare perspective. PharmacoEconomics2004;22:32141.

    [57] Henriksson F. Applications of economic models in healthcare.

    The introduction of Pioglitazone in Sweden. PharmacoEconomics

    2002;20(Suppl 1):4353.

    [58] Coyle D, Palmer AJ, Tam R. Economic evaluation of Pioglitazone

    hydrochloride in the management of type 2 diabetes mellitus in

    Canada. PharmacoEconomics 2002;20(Suppl 1):3142.

    [59] Kalkesar I, Iyer S, Mody R, Rajagopalan R, Kavookjian J. Utilization

    and costs for compliant patients initiating therapy with Pioglitazone or

    Rosiglitazone versus insulin in a Medicaid fee-for-service population.

    J Manag Care Pharm 2006;12:1219.

    [60] Bottomley J, Palmer AJ, Williams R, Dormandy J, Massi-Benedetti M.

    PROactive 03: pioglitazone, type 2 diabetes and reducing macro-

    vascular events economic implications? Br J Diabetes Vasc Dis

    2006;6:6370.[61] Kendall DM. Thiazolidinediones. The case for early use. Diabetes Care

    2006;29:1547.

    [62] Ovalle F, Ovalle-Bermen JF. Thiazolidinediones: a review of their

    benefits and risks. South Med J 2002;95:118894.

    25J.W.F. Elte, J.F. Blickl / European Journal of Internal Medicine 18 (2007) 1825